1
|
Zhang D, Li G, Liu X, Wang Y, Wu J, Ren Y, She G, Zheng D, Zhao Y, Deng XL, Li M, Zhao L. K Ca3.1 upregulation mediated by Ang II-induced JNK/AP-1 activation contributes to atrial fibrosis. Cell Signal 2025; 131:111731. [PMID: 40064281 DOI: 10.1016/j.cellsig.2025.111731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/27/2025] [Accepted: 03/07/2025] [Indexed: 03/15/2025]
Abstract
Atrial fibrillation is strongly associated with an increased risk of embolism, stroke, and heart failure. Current therapeutic approaches often have limited efficacy, and controlling atrial fibrosis remains a critical objective for upstream therapies. The specific mechanisms driving atrial fibrosis remain incompletely understood. The intermediate-conductance calcium-activated potassium channel KCa3.1 has been implicated in promoting fibroblast activation in various fibrotic diseases. This study investigates the role of angiotensin II (Ang II) in regulating KCa3.1, as well as its involvement in the pathogenesis of atrial fibrosis and the underlying signaling mechanisms. In a rat model, chronic Ang II infusion for 4 weeks induced atrial fibrosis, which was significantly attenuated by TRAM-34, a specific KCa3.1 channel blocker. In cultured rat atrial fibroblasts, Ang II treatment promoted fibroblast differentiation, proliferation, migration and collagen production, effects that were suppressed by TRAM-34 and KCa3.1 knockdown. Overexpression of KCa3.1 in fibroblasts further confirmed its pro-fibrotic role. Mechanistically, Ang II upregulated KCa3.1 expression and current density by activating the JNK/AP-1 signaling pathway. This involved phosphorylation of JNK, c-Jun, and c-Fos, leading to the formation of c-Jun/c-Fos heterodimers that directly bound to the KCa3.1 promoter to enhance its transcription. Together, these findings demonstrate that KCa3.1 mediates fibroblast activation and atrial fibrosis through the JNK/AP-1 pathway.
Collapse
Affiliation(s)
- Dongmei Zhang
- Department of Pathology and Pathophysiology, Suzhou Medical College of Soochow University, 199 Ren-ai Road, Suzhou 215123, Jiangsu, China
| | - Guangyao Li
- Department of Pathology and Pathophysiology, Suzhou Medical College of Soochow University, 199 Ren-ai Road, Suzhou 215123, Jiangsu, China
| | - Xiang Liu
- Department of Pathology and Pathophysiology, Suzhou Medical College of Soochow University, 199 Ren-ai Road, Suzhou 215123, Jiangsu, China
| | - Yan Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061, Shaanxi, China
| | - Jie Wu
- Department of Pathology and Pathophysiology, Suzhou Medical College of Soochow University, 199 Ren-ai Road, Suzhou 215123, Jiangsu, China
| | - Yujie Ren
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061, Shaanxi, China
| | - Gang She
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061, Shaanxi, China
| | - Dong Zheng
- Department of Pathology and Pathophysiology, Suzhou Medical College of Soochow University, 199 Ren-ai Road, Suzhou 215123, Jiangsu, China
| | - Yinxia Zhao
- Central Laboratory, Shanghai Xuhui Central Hospital, 366 North Longchuan Road, Shanghai 200031, China
| | - Xiu-Ling Deng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061, Shaanxi, China
| | - Min Li
- Institute of Biology and Medical Sciences, Soochow University, 199 Ren-ai Road, Suzhou 215123, Jiangsu, China.
| | - Limei Zhao
- Department of Pathology and Pathophysiology, Suzhou Medical College of Soochow University, 199 Ren-ai Road, Suzhou 215123, Jiangsu, China; MOE Key Laboratory of Geriatric Diseases and Immunology, School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, Jiangsu, China.
| |
Collapse
|
2
|
Van NTH, Kim WK, Nam JH. Challenges in the Therapeutic Targeting of KCa Channels: From Basic Physiology to Clinical Applications. Int J Mol Sci 2024; 25:2965. [PMID: 38474212 PMCID: PMC10932353 DOI: 10.3390/ijms25052965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 03/14/2024] Open
Abstract
Calcium-activated potassium (KCa) channels are ubiquitously expressed throughout the body and are able to regulate membrane potential and intracellular calcium concentrations, thereby playing key roles in cellular physiology and signal transmission. Consequently, it is unsurprising that KCa channels have been implicated in various diseases, making them potential targets for pharmaceutical interventions. Over the past two decades, numerous studies have been conducted to develop KCa channel-targeting drugs, including those for disorders of the central and peripheral nervous, cardiovascular, and urinary systems and for cancer. In this review, we synthesize recent findings regarding the structure and activating mechanisms of KCa channels. We also discuss the role of KCa channel modulators in therapeutic medicine. Finally, we identify the major reasons behind the delay in bringing these modulators to the pharmaceutical market and propose new strategies to promote their application.
Collapse
Affiliation(s)
- Nhung Thi Hong Van
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea;
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Republic of Korea
| | - Woo Kyung Kim
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Republic of Korea
- Department of Internal Medicine, Graduate School of Medicine, Dongguk University, Goyang 10326, Republic of Korea
| | - Joo Hyun Nam
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea;
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Republic of Korea
| |
Collapse
|
3
|
Takács R, Kovács P, Ebeid RA, Almássy J, Fodor J, Ducza L, Barrett-Jolley R, Lewis R, Matta C. Ca2+-Activated K+ Channels in Progenitor Cells of Musculoskeletal Tissues: A Narrative Review. Int J Mol Sci 2023; 24:ijms24076796. [PMID: 37047767 PMCID: PMC10095002 DOI: 10.3390/ijms24076796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/01/2023] [Accepted: 04/04/2023] [Indexed: 04/08/2023] Open
Abstract
Musculoskeletal disorders represent one of the main causes of disability worldwide, and their prevalence is predicted to increase in the coming decades. Stem cell therapy may be a promising option for the treatment of some of the musculoskeletal diseases. Although significant progress has been made in musculoskeletal stem cell research, osteoarthritis, the most-common musculoskeletal disorder, still lacks curative treatment. To fine-tune stem-cell-based therapy, it is necessary to focus on the underlying biological mechanisms. Ion channels and the bioelectric signals they generate control the proliferation, differentiation, and migration of musculoskeletal progenitor cells. Calcium- and voltage-activated potassium (KCa) channels are key players in cell physiology in cells of the musculoskeletal system. This review article focused on the big conductance (BK) KCa channels. The regulatory function of BK channels requires interactions with diverse sets of proteins that have different functions in tissue-resident stem cells. In this narrative review article, we discuss the main ion channels of musculoskeletal stem cells, with a focus on calcium-dependent potassium channels, especially on the large conductance BK channel. We review their expression and function in progenitor cell proliferation, differentiation, and migration and highlight gaps in current knowledge on their involvement in musculoskeletal diseases.
Collapse
Affiliation(s)
- Roland Takács
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Patrik Kovács
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Rana Abdelsattar Ebeid
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - János Almássy
- Department of Physiology, Faculty of Medicine, Semmelweis University, H-1428 Budapest, Hungary
| | - János Fodor
- Department of Physiology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - László Ducza
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Richard Barrett-Jolley
- Department of Musculoskeletal Biology, Faculty of Health and Life Sciences, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L69 3GA, UK
| | - Rebecca Lewis
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK
| | - Csaba Matta
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| |
Collapse
|
4
|
Dong Y, Suryani L, Zhou X, Muthukumaran P, Rakshit M, Yang F, Wen F, Hassanbhai AM, Parida K, Simon DT, Iandolo D, Lee PS, Ng KW, Teoh SH. Synergistic Effect of PVDF-Coated PCL-TCP Scaffolds and Pulsed Electromagnetic Field on Osteogenesis. Int J Mol Sci 2021; 22:6438. [PMID: 34208563 PMCID: PMC8234164 DOI: 10.3390/ijms22126438] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/02/2021] [Accepted: 06/12/2021] [Indexed: 01/15/2023] Open
Abstract
Bone exhibits piezoelectric properties. Thus, electrical stimulations such as pulsed electromagnetic fields (PEMFs) and stimuli-responsive piezoelectric properties of scaffolds have been investigated separately to evaluate their efficacy in supporting osteogenesis. However, current understanding of cells responding under the combined influence of PEMF and piezoelectric properties in scaffolds is still lacking. Therefore, in this study, we fabricated piezoelectric scaffolds by functionalization of polycaprolactone-tricalcium phosphate (PCL-TCP) films with a polyvinylidene fluoride (PVDF) coating that is self-polarized by a modified breath-figure technique. The osteoinductive properties of these PVDF-coated PCL-TCP films on MC3T3-E1 cells were studied under the stimulation of PEMF. Piezoelectric and ferroelectric characterization demonstrated that scaffolds with piezoelectric coefficient d33 = -1.2 pC/N were obtained at a powder dissolution temperature of 100 °C and coating relative humidity (RH) of 56%. DNA quantification showed that cell proliferation was significantly enhanced by PEMF as low as 0.6 mT and 50 Hz. Hydroxyapatite staining showed that cell mineralization was significantly enhanced by incorporation of PVDF coating. Gene expression study showed that the combination of PEMF and PVDF coating promoted late osteogenic gene expression marker most significantly. Collectively, our results suggest that the synergistic effects of PEMF and piezoelectric scaffolds on osteogenesis provide a promising alternative strategy for electrically augmented osteoinduction. The piezoelectric response of PVDF by PEMF, which could provide mechanical strain, is particularly interesting as it could deliver local mechanical stimulation to osteogenic cells using PEMF.
Collapse
Affiliation(s)
- Yibing Dong
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore; (Y.D.); (X.Z.); (M.R.); (K.P.); (P.S.L.)
| | - Luvita Suryani
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459, Singapore; (L.S.); (P.M.); (F.Y.); (F.W.); (A.M.H.)
| | - Xinran Zhou
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore; (Y.D.); (X.Z.); (M.R.); (K.P.); (P.S.L.)
| | - Padmalosini Muthukumaran
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459, Singapore; (L.S.); (P.M.); (F.Y.); (F.W.); (A.M.H.)
| | - Moumita Rakshit
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore; (Y.D.); (X.Z.); (M.R.); (K.P.); (P.S.L.)
| | - Fengrui Yang
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459, Singapore; (L.S.); (P.M.); (F.Y.); (F.W.); (A.M.H.)
| | - Feng Wen
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459, Singapore; (L.S.); (P.M.); (F.Y.); (F.W.); (A.M.H.)
| | - Ammar Mansoor Hassanbhai
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459, Singapore; (L.S.); (P.M.); (F.Y.); (F.W.); (A.M.H.)
| | - Kaushik Parida
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore; (Y.D.); (X.Z.); (M.R.); (K.P.); (P.S.L.)
| | - Daniel T. Simon
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, 601 74 Norrköping, Sweden; (D.T.S.); (D.I.)
| | - Donata Iandolo
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, 601 74 Norrköping, Sweden; (D.T.S.); (D.I.)
- Mines-Saint-Étienne, Campus Santé Innovations, 10 rue de la Marandière, 42270 Saint-Priest-en-Jarez, France
| | - Pooi See Lee
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore; (Y.D.); (X.Z.); (M.R.); (K.P.); (P.S.L.)
| | - Kee Woei Ng
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore; (Y.D.); (X.Z.); (M.R.); (K.P.); (P.S.L.)
- Center for Nanotechnology and Nanotoxicology, Harvard T.H. Chan School of Public Health, Harvard University, 677 Huntington Avenue, Boston, MA 02115, USA
- Environmental Chemistry and Materials Centre, Nanyang Environment and Water Research Institute, Nanyang Technological University, 1 Cleantech Loop, CleanTech One, Singapore 637141, Singapore
| | - Swee Hin Teoh
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459, Singapore; (L.S.); (P.M.); (F.Y.); (F.W.); (A.M.H.)
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore
| |
Collapse
|
5
|
Matta C, Lewis R, Fellows C, Diszhazi G, Almassy J, Miosge N, Dixon J, Uribe MC, May S, Poliska S, Barrett-Jolley R, Fodor J, Szentesi P, Hajdú T, Keller-Pinter A, Henslee E, Labeed FH, Hughes MP, Mobasheri A. Transcriptome-based screening of ion channels and transporters in a migratory chondroprogenitor cell line isolated from late-stage osteoarthritic cartilage. J Cell Physiol 2021; 236:7421-7439. [PMID: 34008188 DOI: 10.1002/jcp.30413] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 04/28/2021] [Accepted: 05/04/2021] [Indexed: 12/13/2022]
Abstract
Chondrogenic progenitor cells (CPCs) may be used as an alternative source of cells with potentially superior chondrogenic potential compared to mesenchymal stem cells (MSCs), and could be exploited for future regenerative therapies targeting articular cartilage in degenerative diseases such as osteoarthritis (OA). In this study, we hypothesised that CPCs derived from OA cartilage may be characterised by a distinct channelome. First, a global transcriptomic analysis using Affymetrix microarrays was performed. We studied the profiles of those ion channels and transporter families that may be relevant to chondroprogenitor cell physiology. Following validation of the microarray data with quantitative reverse transcription-polymerase chain reaction, we examined the role of calcium-dependent potassium channels in CPCs and observed functional large-conductance calcium-activated potassium (BK) channels involved in the maintenance of the chondroprogenitor phenotype. In line with our very recent results, we found that the KCNMA1 gene was upregulated in CPCs and observed currents that could be attributed to the BK channel. The BK channel inhibitor paxilline significantly inhibited proliferation, increased the expression of the osteogenic transcription factor RUNX2, enhanced the migration parameters, and completely abolished spontaneous Ca2+ events in CPCs. Through characterisation of their channelome we demonstrate that CPCs are a distinct cell population but are highly similar to MSCs in many respects. This study adds key mechanistic data to the in-depth characterisation of CPCs and their phenotype in the context of cartilage regeneration.
Collapse
Affiliation(s)
- Csaba Matta
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Department of Veterinary Preclinical Sciences, School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Rebecca Lewis
- Department of Veterinary Preclinical Sciences, School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Christopher Fellows
- Department of Veterinary Preclinical Sciences, School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Gyula Diszhazi
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Janos Almassy
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Nicolai Miosge
- Department of Prosthodontics, Tissue Regeneration Work Group, Georg August University, Göttingen, Germany
| | - James Dixon
- Wolfson Centre for Stem Cells, Tissue Engineering and Modelling, Centre of Biomolecular Sciences, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Marcos C Uribe
- The Nottingham Arabidopsis Stock Centre (NASC), School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | - Sean May
- The Nottingham Arabidopsis Stock Centre (NASC), School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | - Szilard Poliska
- Genomic Medicine and Bioinformatic Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Richard Barrett-Jolley
- Department of Musculoskeletal Biology, Faculty of Health and Life Sciences, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Janos Fodor
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Peter Szentesi
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tibor Hajdú
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Aniko Keller-Pinter
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Erin Henslee
- Department of Mechanical Engineering Sciences, Centre for Biomedical Engineering, University of Surrey, Guildford, UK
| | - Fatima H Labeed
- Department of Mechanical Engineering Sciences, Centre for Biomedical Engineering, University of Surrey, Guildford, UK
| | - Michael P Hughes
- Department of Mechanical Engineering Sciences, Centre for Biomedical Engineering, University of Surrey, Guildford, UK
| | - Ali Mobasheri
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania.,Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland.,Departments of Orthopedics, Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Joint Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
6
|
Henao JC, Grismaldo A, Barreto A, Rodríguez-Pardo VM, Mejía-Cruz CC, Leal-Garcia E, Pérez-Núñez R, Rojas P, Latorre R, Carvacho I, Torres YP. TRPM8 Channel Promotes the Osteogenic Differentiation in Human Bone Marrow Mesenchymal Stem Cells. Front Cell Dev Biol 2021; 9:592946. [PMID: 33614639 PMCID: PMC7890257 DOI: 10.3389/fcell.2021.592946] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 01/05/2021] [Indexed: 11/29/2022] Open
Abstract
Various families of ion channels have been characterized in mesenchymal stem cells (MSCs), including some members of transient receptor potential (TRP) channels family. TRP channels are involved in critical cellular processes as differentiation and cell proliferation. Here, we analyzed the expression of TRPM8 channel in human bone marrow MSCs (hBM-MSCs), and its relation with osteogenic differentiation. Patch-clamp recordings showed that hBM-MSCs expressed outwardly rectifying currents which were increased by exposure to 500 μM menthol and were partially inhibited by 10 μM of BCTC, a TRPM8 channels antagonist. Additionally, we have found the expression of TRPM8 by RT-PCR and western blot. We also explored the TRPM8 localization in hBM-MSCs by immunofluorescence using confocal microscopy. Remarkably, hBM-MSCs treatment with 100 μM of menthol or 10 μM of icilin, TRPM8 agonists, increases osteogenic differentiation. Conversely, 20 μM of BCTC, induced a decrease of osteogenic differentiation. These results suggest that TRPM8 channels are functionally active in hBM-MSCs and have a role in cell differentiation.
Collapse
Affiliation(s)
- Juan C Henao
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Adriana Grismaldo
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Alfonso Barreto
- Grupo de Inmunobiología y Biología Celular, Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Viviana M Rodríguez-Pardo
- Grupo de Inmunobiología y Biología Celular, Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Claudia Camila Mejía-Cruz
- Grupo de Inmunobiología y Biología Celular, Departamento de Microbiología, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Efrain Leal-Garcia
- Departamento de Ortopedia y Traumatología, Facultad de Medicina, Pontificia Universidad Javeriana, Bogotá, Colombia
| | | | - Patricio Rojas
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Ramón Latorre
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Ingrid Carvacho
- Department of Biology and Chemistry, Faculty of Basic Sciences, Universidad Católica del Maule, Talca, Chile
| | - Yolima P Torres
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| |
Collapse
|
7
|
Naghibzadeh M, Gholampour S, Naghibzadeh M, Sadeghian-Nodoushan F, Nikukar H. The effect of electromagnetic field on decreasing and increasing of the growth and proliferation rate of dermal fibroblast cell. Dermatol Ther 2020; 33:e13803. [PMID: 32526050 DOI: 10.1111/dth.13803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 04/06/2020] [Accepted: 06/06/2020] [Indexed: 11/29/2022]
Abstract
Maintaining the health of dermal fibroblast cells and controlling their growth and proliferation would directly affect the health of skin tissues. The present study encompassed three control and three experimental specimens, which were different in terms of the duration of exposure to electromagnetic field (EMF) and intensity. With a decrease in intensity from 2 to 1 mT during 24, 48, and 72 h after exposing the cells to EMF, the frequency of the sample fibroblast cells increased by 60.3%, 144.9%, and 90.1%, respectively. With an increase in intensity from 3 to 4 mT during 48 and 72 h of exposure to EMF, the frequencies of the sample fibroblast cells decreased by 6.8% and 86.7%, respectively. It seems to be possible to achieve the most desirable condition to help the restoration of wounds and skin lesions through decreasing the exposure intensity from 2 to 0.5 mT and increasing EMF exposure time from 24 to 72 h simultaneously and non-invasively. The most desirable approach to improve the treatment of skin cancers non-invasively is to increase the intensity from 3 to 5 mT and to enhance EMF exposure time from 48 to 72 h.
Collapse
Affiliation(s)
- Mehran Naghibzadeh
- Department of Biomedical Engineering, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Seifollah Gholampour
- Department of Biomedical Engineering, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Majid Naghibzadeh
- Medical Nanotechnology and Tissue Engineering Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Sadeghian-Nodoushan
- Medical Nanotechnology and Tissue Engineering Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Department of Tissue engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Habib Nikukar
- Medical Nanotechnology and Tissue Engineering Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
8
|
Jia X, Su H, Chen X, Huang Y, Zheng Y, Ji P, Gao C, Gong X, Huang Y, Jiang LH, Fan Y. A critical role of the K Ca 3.1 channel in mechanical stretch-induced proliferation of rat bone marrow-derived mesenchymal stem cells. J Cell Mol Med 2020; 24:3739-3744. [PMID: 32065503 PMCID: PMC7131943 DOI: 10.1111/jcmm.15014] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/02/2020] [Accepted: 01/06/2020] [Indexed: 12/11/2022] Open
Abstract
Mechanical stimulation is an important factor regulating mesenchymal stem cell (MSC) functions such as proliferation. The Ca2+ -activated K+ channel, KCa 3.1, is critically engaged in MSC proliferation but its role in mechanical regulation of MSC proliferation remains unknown. Here, we examined the KCa 3.1 channel expression and its role in rat bone marrow-derived MSC (BMSC) proliferation in response to mechanical stretch. Application of mechanical stretch stimulated BMSC proliferation via promoting cell cycle progression. Such mechanical stimulation up-regulated the KCa 3.1 channel expression and pharmacological or genetic inhibition of the KCa 3.1 channel strongly suppressed stretch-induced increase in cell proliferation and cell cycle progression. These results support that the KCa 3.1 channel plays an important role in transducing mechanical forces to MSC proliferation. Our finding provides new mechanistic insights into how mechanical stimuli regulate MSC proliferation and also a viable bioengineering approach to improve MSC proliferation.
Collapse
Affiliation(s)
- Xiaoling Jia
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China.,Beijing Advanced Innovation Centre for Biomedical Engineering, Beihang University, Beijing, China.,School of Biomedical Science, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Hao Su
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Xinlan Chen
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Yangbi Huang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Yufan Zheng
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Pei Ji
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Chao Gao
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Xianghui Gong
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Yan Huang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Lin-Hua Jiang
- School of Biomedical Science, Faculty of Biological Sciences, University of Leeds, Leeds, UK.,Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China.,Beijing Advanced Innovation Centre for Biomedical Engineering, Beihang University, Beijing, China.,Beijing Key Laboratory of Rehabilitation Technical Aids for Old-Age Disability, National Research Center for Rehabilitation Technical Aids, Beijing, China
| |
Collapse
|
9
|
Aldebs AI, Zohora FT, Nosoudi N, Singh SP, Ramirez‐Vick JE. Effect of Pulsed Electromagnetic Fields on Human Mesenchymal Stem Cells Using 3D Magnetic Scaffolds. Bioelectromagnetics 2020; 41:175-187. [PMID: 31944364 PMCID: PMC9290550 DOI: 10.1002/bem.22248] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 01/01/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Alyaa I. Aldebs
- Department of Biomedical, Industrial & Human Factors EngineeringWright State UniversityDayton Ohio
| | - Fatema T. Zohora
- Department of Biomedical, Industrial & Human Factors EngineeringWright State UniversityDayton Ohio
| | - Nasim Nosoudi
- Biomedical Engineering ProgramMarshall UniversityHuntington West Virginia
| | | | - Jaime E. Ramirez‐Vick
- Department of Biomedical, Industrial & Human Factors EngineeringWright State UniversityDayton Ohio
| |
Collapse
|
10
|
Hu XM, Ren S, Li K, Li XT. Tacrine modulates Kv2.1 channel gene expression and cell proliferation. Int J Neurosci 2020; 130:781-787. [PMID: 31847645 DOI: 10.1080/00207454.2019.1705811] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Purpose/Aim: Besides as a cholinesterase (ChE) inhibitor, tacrine is able to act on multiple targets such as nicotinic receptors (nAChRs) and voltage-gated K+ (Kv) channels. Kv2.1, a Kv channel subunit underlying delayed rectifier currents with slow kinetics of inactivation, is highly expressed in the mammalian brain, especially in the hippocampus. Nevertheless, limited data are available concerning the relationship between tacrine and Kv2.1 channels. In the present study, we explore the possible effects of tacrine on Kv2.1 channels in heterologous expression systems and N2A cells.Materials and methods: The change of expression and currents of Kv2.1 after treatment with tacrine was detected by PCR and whole-cell recordings, respectively. WST-8 experiments were performed to reveal the effects of tacrine on cell proliferation.Results: Incubation with tacrine induced a significant reduction of the mRNA level of Kv2.1 channels in HEK293 cells. The decline of corresponding currents carried by Kv2.1 was also observed. Moreover, the proliferation rates of HEK293 cells with Kv2.1 channel were substantially enhanced after treatment with this chemical for 24 h. Similar results were also detected after exposure to tacrine in N2A cells with native expression of Kv2.1 channels.Conclusion: These lines of evidence indicate that application of tacrine downregulates the expression of Kv2.1 channels and increase cell proliferation. The effect of tacrine on Kv2.1 channels may provide an alternative explanation for its neuroprotective action.
Collapse
Affiliation(s)
- Xi-Mu Hu
- Graduate Institute of South-Central University for Nationalities, Wuhan, China
| | - Sheng Ren
- College of Life Science, South-Central University for Nationalities, Wuhan, China
| | - Kai Li
- Graduate Institute of South-Central University for Nationalities, Wuhan, China
| | - Xian-Tao Li
- College of Life Science, South-Central University for Nationalities, Wuhan, China
| |
Collapse
|
11
|
She G, Ren YJ, Wang Y, Hou MC, Wang HF, Gou W, Lai BC, Lei T, Du XJ, Deng XL. K Ca3.1 Channels Promote Cardiac Fibrosis Through Mediating Inflammation and Differentiation of Monocytes Into Myofibroblasts in Angiotensin II -Treated Rats. J Am Heart Assoc 2020; 8:e010418. [PMID: 30563389 PMCID: PMC6405723 DOI: 10.1161/jaha.118.010418] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background Cardiac fibrosis is a core pathological process associated with heart failure. The recruitment and differentiation of primitive fibroblast precursor cells of bone marrow origin play a critical role in pathological interstitial cardiac fibrosis. The KC a3.1 channels are expressed in both ventricular fibroblasts and circulating mononuclear cells in rats and are upregulated by angiotensin II . We hypothesized that KC a3.1 channels mediate the inflammatory microenvironment in the heart, promoting the infiltrated bone marrow-derived circulating mononuclear cells to differentiate into myofibroblasts, leading to myocardial fibrosis. Methods and Results We established a cardiac fibrosis model in rats by infusing angiotensin II to evaluate the impact of the specific KC a3.1 channel blocker TRAM -34 on cardiac fibrosis. At the same time, mouse CD 4+ T cells and rat circulating mononuclear cells were separated to investigate the underlying mechanism of the TRAM -34 anti-cardiac fibrosis effect. TRAM -34 significantly attenuated cardiac fibrosis and the inflammatory reaction and reduced the number of fibroblast precursor cells and myofibroblasts. Inhibition of KC a3.1 channels suppressed angiotensin II -stimulated expression and secretion of interleukin-4 and interleukin-13 in CD 4+ T cells and interleukin-4- or interleukin-13-induced differentiation of monocytes into fibrocytes. Conclusions KC a3.1 channels facilitate myocardial inflammation and the differentiation of bone marrow-derived monocytes into myofibroblasts in cardiac fibrosis caused by angiotensin II infusion.
Collapse
Affiliation(s)
- Gang She
- 1 Department of Physiology and Pathophysiology School of Basic Medical Sciences Xi'an Jiaotong University Health Science Center Xi'an Shaanxi China
| | - Yu-Jie Ren
- 1 Department of Physiology and Pathophysiology School of Basic Medical Sciences Xi'an Jiaotong University Health Science Center Xi'an Shaanxi China.,5 Department of Pathology Xi'an Guangren Hospital Affiliated to Xi'an Jiaotong University Health Science Center Xi'an Shaanxi China
| | - Yan Wang
- 1 Department of Physiology and Pathophysiology School of Basic Medical Sciences Xi'an Jiaotong University Health Science Center Xi'an Shaanxi China
| | - Meng-Chen Hou
- 1 Department of Physiology and Pathophysiology School of Basic Medical Sciences Xi'an Jiaotong University Health Science Center Xi'an Shaanxi China
| | - Hui-Fang Wang
- 5 Department of Pathology Xi'an Guangren Hospital Affiliated to Xi'an Jiaotong University Health Science Center Xi'an Shaanxi China
| | - Wei Gou
- 3 Basic Experiment Teaching Center School of Basic Medical Sciences Xi'an Jiaotong University Health Science Center Xi'an Shaanxi China
| | - Bao-Chang Lai
- 4 Cardiovascular Research Centre School of Basic Medical Sciences Xi'an Jiaotong University Health Science Center Xi'an Shaanxi China
| | - Ting Lei
- 2 Department of Pathology School of Basic Medical Sciences Xi'an Jiaotong University Health Science Center Xi'an Shaanxi China
| | - Xiao-Jun Du
- 1 Department of Physiology and Pathophysiology School of Basic Medical Sciences Xi'an Jiaotong University Health Science Center Xi'an Shaanxi China.,6 Baker Heart and Diabetes Institute Melbourne Victoria Australia
| | - Xiu-Ling Deng
- 1 Department of Physiology and Pathophysiology School of Basic Medical Sciences Xi'an Jiaotong University Health Science Center Xi'an Shaanxi China.,4 Cardiovascular Research Centre School of Basic Medical Sciences Xi'an Jiaotong University Health Science Center Xi'an Shaanxi China
| |
Collapse
|
12
|
Xu Y, Wang Y, Pang X, Li Z, Wu J, Zhou Z, Xu T, Gobin Beharee R, Jin L, Yu J. Potassium dihydrogen phosphate promotes the proliferation and differentiation of human periodontal ligament stem cells via nuclear factor kappa B pathway. Exp Cell Res 2019; 384:111593. [PMID: 31487508 DOI: 10.1016/j.yexcr.2019.111593] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 08/28/2019] [Accepted: 08/31/2019] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Periodontal ligament stem cells (PDLSCs) are vital for the regeneration of periodontal tissues. Potassium dihydrogen phosphate (KH2PO4) has recently been applied as a component of the mineralization inducing medium (MM), which can be used to induce osteogenic differentiation of dental stem cells. However, whether KH2PO4 has effects on PDLSCs has not been studied. MATERIALS AND METHODS PDLSCs were isolated by magnetic activated cell sorting and cultured. Alkaline phosphatase (ALP) activity and ALP protein expression of PDLSCs treated with different concentrations of KH2PO4 were examined to make sure the optimal concentration of KH2PO4 for the following experiments. The effects of KH2PO4 on the proliferation and differentiation of PDLSCs were investigated by flow cytometry, cell counting kit-8 assay, alizarin red staining, real-time RT-PCR, and Western blot. The involvement of nuclear factor kappa B (NF-κB) pathway in KH2PO4-treated PDLSCs was analyzed by Western blot and alizarin red staining. RESULTS ALP activity assay and ALP protein expression examination revealed that 1.8 mmol/L KH2PO4 was the optimal concentration for the induction of hPDLSCs by KH2PO4. The proliferation and mineralization capacity of PDLSCs treated with KH2PO4 were enhanced as compared with the control group. PDLSCs treated with KH2PO4 showed an improved proliferation capacity in logarithmic growth phase at day 7. As PDLSCs were treated with KH2PO4, the expression of odonto/osteogenic markers (OCN/OCN, DSP/DSPP, OSX/OSX, RUNX2/RUNX2, and ALP/ALP) in cells were up-regulated at day 3 or 7. Moreover, the expression of IκBα in cytoplasm was down-regulated, along with an increased expression of p-P65 in cytoplasm and an up-regulated expression of P65 in nucleus. When treated with BMS345541 (the specific NF-κB inhibitor), the odonto/osteogenic differentiation of KH2PO4-treated PDLSCs was significantly attenuated. CONCLUSION KH2PO4 can improve the proliferation and odonto/osteogenic differentiation capacity of PDLSCs via NF-κB pathway, and thus represents a potential target involved in the regeneration of periodontium for clinical treatments.
Collapse
Affiliation(s)
- Yunlong Xu
- Endodontic Department, Changzhou Stomatological Hospital, 61 Beizhi Street, Changzhou, Jiangsu 213000, China; Key Laboratory of Oral Diseases of Jiangsu Province and Stomatological Institute of Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu 210029, China
| | - Yanqiu Wang
- Key Laboratory of Oral Diseases of Jiangsu Province and Stomatological Institute of Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu 210029, China; Endodontic Department, School of Stomatology, Nanjing Medical University, 136 Hanzhong Road, Nanjing, Jiangsu 210029, China
| | - Xiyao Pang
- Key Laboratory of Oral Diseases of Jiangsu Province and Stomatological Institute of Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu 210029, China; Endodontic Department, School of Stomatology, Nanjing Medical University, 136 Hanzhong Road, Nanjing, Jiangsu 210029, China
| | - Zehan Li
- Key Laboratory of Oral Diseases of Jiangsu Province and Stomatological Institute of Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu 210029, China; Endodontic Department, School of Stomatology, Nanjing Medical University, 136 Hanzhong Road, Nanjing, Jiangsu 210029, China
| | - Jintao Wu
- Key Laboratory of Oral Diseases of Jiangsu Province and Stomatological Institute of Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu 210029, China; Endodontic Department, School of Stomatology, Nanjing Medical University, 136 Hanzhong Road, Nanjing, Jiangsu 210029, China
| | - Zhou Zhou
- Key Laboratory of Oral Diseases of Jiangsu Province and Stomatological Institute of Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu 210029, China; Endodontic Department, School of Stomatology, Nanjing Medical University, 136 Hanzhong Road, Nanjing, Jiangsu 210029, China
| | - Tao Xu
- Key Laboratory of Oral Diseases of Jiangsu Province and Stomatological Institute of Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu 210029, China
| | - Romila Gobin Beharee
- Key Laboratory of Oral Diseases of Jiangsu Province and Stomatological Institute of Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu 210029, China; Endodontic Department, School of Stomatology, Nanjing Medical University, 136 Hanzhong Road, Nanjing, Jiangsu 210029, China
| | - Lin Jin
- Key Laboratory of Oral Diseases of Jiangsu Province and Stomatological Institute of Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu 210029, China; Nantong Stomatological Hospital, 36 South Yuelong Road, Nantong, Jiangsu 226001, China
| | - Jinhua Yu
- Key Laboratory of Oral Diseases of Jiangsu Province and Stomatological Institute of Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu 210029, China; Endodontic Department, School of Stomatology, Nanjing Medical University, 136 Hanzhong Road, Nanjing, Jiangsu 210029, China.
| |
Collapse
|
13
|
Silva HB, Rodrigues DC, Andrade R, Teixeira GHGSF, Stelling MP, Ponte CG, Nascimento JHM, Campos de Carvalho AC, Medei E. Expression of potassium channels is relevant for cell survival and migration in a murine bone marrow stromal cell line. J Cell Physiol 2019; 234:18086-18097. [PMID: 30887515 DOI: 10.1002/jcp.28441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 02/07/2019] [Accepted: 02/14/2019] [Indexed: 02/05/2023]
Abstract
S17 is a clonogenic bone marrow stromal (BMS) cell line derived from mouse that has been extensively used to assess both human and murine hematopoiesis support capacity. However, very little is known about the expression of potassium ion channels and their function in cell survival and migration in these cells. Thus, the present study was designed to characterize potassium ion channels using electrophysiological and molecular biological approaches in S17 BMS cells. The whole-cell configuration of the patch clamp technique has been applied to identify potassium ion currents and reverse transcription polymerase chain reaction (RT-PCR) used to determine their molecular identities. Based on gating kinetics and pharmacological modulation of the macroscopic currents we found the presence of four functional potassium ion channels in S17 BMS cells. These include a current rapidly activated and inactivated, tetraethylammonium-sensitive, (IKV ) in most (50%) cells; a fast activated and rapidly inactivating A-type K + current (IK A -like); a delayed rectifier K + current (IK DR ) and an inward rectifier potassium current (IK IR ), found in, respectively 4.5%, 26% and 24% of these cells. RT-PCR confirmed the presence of mRNA transcripts for the alpha subunit of the corresponding functional ion channels. Additionally, functional assays were performed to investigate the importance of potassium currents in cell survival and migration. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide analyses revealed a reduction in cell viability, while wound healing assays revealed reduced migration potential in cells incubated with different potassium channel blockers. In conclusion, our data suggested that potassium currents might play a role in the maintenance of overall S17 cell ionic homeostasis directly affecting cell survival and migration.
Collapse
Affiliation(s)
- Henrique B Silva
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Center for Neuroscience and Cell Biology, University of Coimbra Rua Larga, Faculty of Medicine, Coimbra, Portugal
| | - Deivid C Rodrigues
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Raiana Andrade
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Gabriel H G S F Teixeira
- Federal Institute of Education, Science and Technology of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Mariana P Stelling
- Federal Institute of Education, Science and Technology of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Cristiano G Ponte
- Federal Institute of Education, Science and Technology of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - José H M Nascimento
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Emiliano Medei
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
14
|
Chubinskiy-Nadezhdin VI, Sudarikova AV, Shilina MA, Vasileva VY, Grinchuk TM, Lyublinskaya OG, Nikolsky NN, Negulyaev YA. Cell Cycle-Dependent Expression of Bk Channels in Human Mesenchymal Endometrial Stem Cells. Sci Rep 2019; 9:4595. [PMID: 30872711 PMCID: PMC6418245 DOI: 10.1038/s41598-019-41096-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Accepted: 02/28/2019] [Indexed: 12/21/2022] Open
Abstract
The study of ion channels in stem cells provides important information about their role in stem cell fate. Previously we have identified the activity of calcium-activated potassium channels of big conductance (BK channels) in human endometrium-derived mesenchymal stem cells (eMSCs). BK channels could have significant impact into signaling processes by modulating membrane potential. The membrane potential and ionic permeability dynamically changes during cycle transitions. Here, we aimed at verification of the role of BK channels as potassium transporting pathway regulating cell cycle passageway of eMSCs. The functional expression of native BK channels was confirmed by patch-clamp and immunocytochemistry. In non-synchronized cells immunofluorescent analysis revealed BK-positive and BK-negative stained eMSCs. Using cell synchronization, we found that the presence of BK channels in plasma membrane was cell cycle-dependent and significantly decreased in G2M phase. However, the study of cell cycle progression in presence of selective BK channel inhibitors showed no effect of pore blockers on cycle transitions. Thus, BK channel-mediated K+ transport is not critical for the fundamental mechanism of passageway through cell cycle of eMSCs. At the same time, the dynamics of the presence of BK channels on plasma membrane of eMSCs can be a novel indicator of cellular proliferation.
Collapse
Affiliation(s)
| | | | - Mariia A Shilina
- Institute of Cytology RAS, 194064, Tikhoretsky Ave. 4, St. Petersburg, Russia
| | - Valeria Y Vasileva
- Institute of Cytology RAS, 194064, Tikhoretsky Ave. 4, St. Petersburg, Russia
| | - Tatiana M Grinchuk
- Institute of Cytology RAS, 194064, Tikhoretsky Ave. 4, St. Petersburg, Russia
| | - Olga G Lyublinskaya
- Institute of Cytology RAS, 194064, Tikhoretsky Ave. 4, St. Petersburg, Russia
| | - Nikolai N Nikolsky
- Institute of Cytology RAS, 194064, Tikhoretsky Ave. 4, St. Petersburg, Russia
| | - Yuri A Negulyaev
- Institute of Cytology RAS, 194064, Tikhoretsky Ave. 4, St. Petersburg, Russia
- Department of Medical Physics, Peter the Great St. Petersburg Polytechnic University, 29, Polytechnicheskaya st., 195251, St. Petersburg, Russia
| |
Collapse
|
15
|
Implication of Voltage-Gated Potassium Channels in Neoplastic Cell Proliferation. Cancers (Basel) 2019; 11:cancers11030287. [PMID: 30823672 PMCID: PMC6468671 DOI: 10.3390/cancers11030287] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 02/21/2019] [Accepted: 02/24/2019] [Indexed: 12/12/2022] Open
Abstract
Voltage-gated potassium channels (Kv) are the largest group of ion channels. Kv are involved in controlling the resting potential and action potential duration in the heart and brain. Additionally, these proteins participate in cell cycle progression as well as in several other important features in mammalian cell physiology, such as activation, differentiation, apoptosis, and cell volume control. Therefore, Kv remarkably participate in the cell function by balancing responses. The implication of Kv in physiological and pathophysiological cell growth is the subject of study, as Kv are proposed as therapeutic targets for tumor regression. Though it is widely accepted that Kv channels control proliferation by allowing cell cycle progression, their role is controversial. Kv expression is altered in many cancers, and their participation, as well as their use as tumor markers, is worthy of effort. There is an ever-growing list of Kv that remodel during tumorigenesis. This review focuses on the actual knowledge of Kv channel expression and their relationship with neoplastic proliferation. In this work, we provide an update of what is currently known about these proteins, thereby paving the way for a more precise understanding of the participation of Kv during cancer development.
Collapse
|
16
|
Marakhova I, Domnina A, Shatrova A, Borodkina A, Burova E, Pugovkina N, Zemelko V, Nikolsky N. Proliferation-related changes in K + content in human mesenchymal stem cells. Sci Rep 2019; 9:346. [PMID: 30674973 PMCID: PMC6344592 DOI: 10.1038/s41598-018-36922-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 11/27/2018] [Indexed: 12/23/2022] Open
Abstract
Intracellular monovalent ions have been shown to be important for cell proliferation, however, mechanisms through which ions regulate cell proliferation is not well understood. Ion transporters may be implicated in the intracellular signaling: Na+ and Cl− participate in regulation of intracellular pH, transmembrane potential, Ca2+ homeostasis. Recently, it is has been suggested that K+ may be involved in “the pluripotency signaling network”. Our study has been focused on the relations between K+ transport and stem cell proliferation. We compared monovalent cation transport in human mesenchymal stem cells (hMSCs) at different passages and at low and high densities of culture as well as during stress-induced cell cycle arrest and revealed a decline in K+ content per cell protein which was associated with accumulation of G1 cells in population and accompanied cell proliferation slowing. It is suggested that cell K+ may be important for successful cell proliferation as the main intracellular ion that participates in regulation of cell volume during cell cycle progression. It is proposed that cell K+ content as related to cell protein is a physiological marker of stem cell proliferation and may be used as an informative test for assessing the functional status of stem cells in vitro.
Collapse
Affiliation(s)
- Irina Marakhova
- Department of Intracellular Signaling and Transport, Institute of Cytology, Academy of Sciences, St-Petersburg, 194064, Russian Federation.
| | - Alisa Domnina
- Department of Intracellular Signaling and Transport, Institute of Cytology, Academy of Sciences, St-Petersburg, 194064, Russian Federation
| | - Alla Shatrova
- Department of Intracellular Signaling and Transport, Institute of Cytology, Academy of Sciences, St-Petersburg, 194064, Russian Federation
| | - Aleksandra Borodkina
- Department of Intracellular Signaling and Transport, Institute of Cytology, Academy of Sciences, St-Petersburg, 194064, Russian Federation
| | - Elena Burova
- Department of Intracellular Signaling and Transport, Institute of Cytology, Academy of Sciences, St-Petersburg, 194064, Russian Federation
| | - Natalja Pugovkina
- Department of Intracellular Signaling and Transport, Institute of Cytology, Academy of Sciences, St-Petersburg, 194064, Russian Federation
| | - Victoria Zemelko
- Department of Intracellular Signaling and Transport, Institute of Cytology, Academy of Sciences, St-Petersburg, 194064, Russian Federation
| | - Nikolay Nikolsky
- Department of Intracellular Signaling and Transport, Institute of Cytology, Academy of Sciences, St-Petersburg, 194064, Russian Federation
| |
Collapse
|
17
|
Polo-Corrales L, Ramirez-Vick J, Feria-Diaz JJ. Recent Advances in Biophysical stimulation of MSC for bone regeneration. ACTA ACUST UNITED AC 2018. [DOI: 10.17485/ijst/2018/v11i15/121405] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
|
18
|
McLaughlin KA, Levin M. Bioelectric signaling in regeneration: Mechanisms of ionic controls of growth and form. Dev Biol 2018; 433:177-189. [PMID: 29291972 PMCID: PMC5753428 DOI: 10.1016/j.ydbio.2017.08.032] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 08/23/2017] [Accepted: 08/28/2017] [Indexed: 12/11/2022]
Abstract
The ability to control pattern formation is critical for the both the embryonic development of complex structures as well as for the regeneration/repair of damaged or missing tissues and organs. In addition to chemical gradients and gene regulatory networks, endogenous ion flows are key regulators of cell behavior. Not only do bioelectric cues provide information needed for the initial development of structures, they also enable the robust restoration of normal pattern after injury. In order to expand our basic understanding of morphogenetic processes responsible for the repair of complex anatomy, we need to identify the roles of endogenous voltage gradients, ion flows, and electric fields. In complement to the current focus on molecular genetics, decoding the information transduced by bioelectric cues enhances our knowledge of the dynamic control of growth and pattern formation. Recent advances in science and technology place us in an exciting time to elucidate the interplay between molecular-genetic inputs and important biophysical cues that direct the creation of tissues and organs. Moving forward, these new insights enable additional approaches to direct cell behavior and may result in profound advances in augmentation of regenerative capacity.
Collapse
Affiliation(s)
- Kelly A McLaughlin
- Allen Discovery Center, Department of Biology, Tufts University, 200 Boston Ave., Suite 4700, Medford, MA 02155, United States.
| | - Michael Levin
- Allen Discovery Center, Department of Biology, Tufts University, 200 Boston Ave., Suite 4700, Medford, MA 02155, United States
| |
Collapse
|
19
|
Ayad O, Magaud C, Sebille S, Bescond J, Mimbimi C, Cognard C, Faivre JF, Bois P, Chatelier A. Functional BKCa channel in human resident cardiac stem cells expressing W8B2. FEBS J 2017; 285:518-530. [PMID: 29211342 DOI: 10.1111/febs.14352] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 11/02/2017] [Accepted: 11/30/2017] [Indexed: 01/24/2023]
Abstract
Recently, a new population of resident cardiac stem cells (CSCs) positive for the W8B2 marker has been identified. These CSCs are considered to be an ideal cellular source to repair myocardial damage after infarction. However, the electrophysiological profile of these cells has not been characterized yet. We first establish the conditions of isolation and expansion of W8B2+ CSCs from human heart biopsies using a magnetic sorting system followed by flow cytometry cell sorting. These cells display a spindle-shaped morphology, are highly proliferative, and possess self-renewal capacity demonstrated by their ability to form colonies. Besides, W8B2+ CSCs are positive for mesenchymal markers but negative for hematopoietic and endothelial ones. RT-qPCR and immunostaining experiments show that W8B2+ CSCs express some early cardiac-specific transcription factors but lack the expression of cardiac-specific structural genes. Using patch clamp in the whole-cell configuration, we show for the first time the electrophysiological signature of BKCa current in these cells. Accordingly, RT-PCR and western blotting analysis confirmed the presence of BKCa at both mRNA and protein levels in W8B2+ CSCs. Interestingly, BKCa channel inhibition by paxilline decreased cell proliferation in a concentration-dependent manner and halted cell cycle progression at the G0/G1 phase. The inhibition of BKCa also decreased the self-renewal capacity but did not affect migration of W8B2+ CSCs. Taken together, our results are consistent with an important role of BKCa channels in cell cycle progression and self-renewal in human cardiac stem cells.
Collapse
Affiliation(s)
- Oualid Ayad
- Equipe Transferts Ioniques et Rythmicité Cardiaque, Laboratoire Signalisation et Transports Ioniques Membranaires, CNRS ERL 7368, EA 7349, Université de Poitiers, France
| | - Christophe Magaud
- Equipe Transferts Ioniques et Rythmicité Cardiaque, Laboratoire Signalisation et Transports Ioniques Membranaires, CNRS ERL 7368, EA 7349, Université de Poitiers, France
| | - Stéphane Sebille
- Equipe Transferts Ioniques et Rythmicité Cardiaque, Laboratoire Signalisation et Transports Ioniques Membranaires, CNRS ERL 7368, EA 7349, Université de Poitiers, France
| | - Jocelyn Bescond
- Equipe Transferts Ioniques et Rythmicité Cardiaque, Laboratoire Signalisation et Transports Ioniques Membranaires, CNRS ERL 7368, EA 7349, Université de Poitiers, France
| | - Chloé Mimbimi
- Service de chirurgie cardio-thoracique, CHU Poitiers, France
| | - Christian Cognard
- Equipe Transferts Ioniques et Rythmicité Cardiaque, Laboratoire Signalisation et Transports Ioniques Membranaires, CNRS ERL 7368, EA 7349, Université de Poitiers, France
| | - Jean-Francois Faivre
- Equipe Transferts Ioniques et Rythmicité Cardiaque, Laboratoire Signalisation et Transports Ioniques Membranaires, CNRS ERL 7368, EA 7349, Université de Poitiers, France
| | - Patrick Bois
- Equipe Transferts Ioniques et Rythmicité Cardiaque, Laboratoire Signalisation et Transports Ioniques Membranaires, CNRS ERL 7368, EA 7349, Université de Poitiers, France
| | - Aurelien Chatelier
- Equipe Transferts Ioniques et Rythmicité Cardiaque, Laboratoire Signalisation et Transports Ioniques Membranaires, CNRS ERL 7368, EA 7349, Université de Poitiers, France
| |
Collapse
|
20
|
Pchelintseva E, Djamgoz MBA. Mesenchymal stem cell differentiation: Control by calcium-activated potassium channels. J Cell Physiol 2017; 233:3755-3768. [PMID: 28776687 DOI: 10.1002/jcp.26120] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 08/01/2017] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) are widely used in modern medicine for which understanding the mechanisms controlling their differentiation is fundamental. Ion channels offer novel insights to this process because of their role in modulating membrane potential and intracellular milieu. Here, we evaluate the contribution of calcium-activated potassium (KCa ) channels to the three main components of MSC differentiation: initiation, proliferation, and migration. First, we demonstrate the importance of the membrane potential (Vm ) and the apparent association of hyperpolarization with differentiation. Of KCa subtypes, most evidence points to activity of big-conductance channels in inducing initiation. On the other hand, intermediate-conductance currents have been shown to promote progression through the cell cycle. While there is no information on the role of KCa channels in migration of MSCs, work from other stem cells and cancer cells suggest that intermediate-conductance and to a lesser extent big-conductance channels drive migration. In all cases, these effects depend on species, tissue origin and lineage. Finally, we present a conceptual model that demonstrates how KCa activity could influence differentiation by regulating Vm and intracellular Ca2+ oscillations. We conclude that KCa channels have significant involvement in MSC differentiation and could potentially enable novel tissue engineering approaches and therapies.
Collapse
Affiliation(s)
- Ekaterina Pchelintseva
- Department of Life Sciences, Imperial College London, South Kensington Campus, Neuroscience Solution to Cancer Research Group, London, UK.,Department of Bioengineering, Imperial College London, South Kensington Campus, London, UK
| | - Mustafa B A Djamgoz
- Department of Life Sciences, Imperial College London, South Kensington Campus, Neuroscience Solution to Cancer Research Group, London, UK
| |
Collapse
|
21
|
Franklin BM, Voss SR, Osborn JL. Ion channel signaling influences cellular proliferation and phagocyte activity during axolotl tail regeneration. Mech Dev 2017; 146:42-54. [PMID: 28603004 PMCID: PMC6386162 DOI: 10.1016/j.mod.2017.06.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 05/24/2017] [Accepted: 06/01/2017] [Indexed: 01/30/2023]
Abstract
Little is known about the potential for ion channels to regulate cellular behaviors during tissue regeneration. Here, we utilized an amphibian tail regeneration assay coupled with a chemical genetic screen to identify ion channel antagonists that altered critical cellular processes during regeneration. Inhibition of multiple ion channels either partially (anoctamin1/Tmem16a, anoctamin2/Tmem16b, KV2.1, KV2.2, L-type CaV channels and H/K ATPases) or completely (GlyR, GABAAR, KV1.5 and SERCA pumps) inhibited tail regeneration. Partial inhibition of tail regeneration by blocking the calcium activated chloride channels, anoctamin1&2, was associated with a reduction of cellular proliferation in tail muscle and mesenchymal regions. Inhibition of anoctamin 1/2 also altered the post-amputation transcriptional response of p44/42 MAPK signaling pathway genes, including decreased expression of erk1/erk2. We also found that complete inhibition via voltage gated K+ channel blockade was associated with diminished phagocyte recruitment to the amputation site. The identification of H+ pumps as required for axolotl tail regeneration supports findings in Xenopus and Planaria models, and more generally, the conservation of ion channels as regulators of tissue regeneration. This study provides a preliminary framework for an in-depth investigation of the mechanistic role of ion channels and their potential involvement in regulating cellular proliferation and other processes essential to wound healing, appendage regeneration, and tissue repair.
Collapse
Affiliation(s)
- Brandon M Franklin
- Department of Biology, University of Kentucky, Lexington, KY 40506, United States
| | - S Randal Voss
- Department of Biology, University of Kentucky, Lexington, KY 40506, United States
| | - Jeffrey L Osborn
- Department of Biology, University of Kentucky, Lexington, KY 40506, United States.
| |
Collapse
|
22
|
Shen H, Bocksteins E, Kondrychyn I, Snyders D, Korzh V. Functional antagonism of voltage-gated K+ channel α-subunits in the developing brain ventricular system. Development 2016; 143:4249-4260. [PMID: 27729411 DOI: 10.1242/dev.140467] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 09/30/2016] [Indexed: 01/10/2023]
Abstract
The brain ventricular system is essential for neurogenesis and brain homeostasis. Its neuroepithelial lining effects these functions, but the underlying molecular pathways remain to be understood. We found that the potassium channels expressed in neuroepithelial cells determine the formation of the ventricular system. The phenotype of a novel zebrafish mutant characterized by denudation of neuroepithelial lining of the ventricular system and hydrocephalus is mechanistically linked to Kcng4b, a homologue of the 'silent' voltage-gated potassium channel α-subunit Kv6.4. We demonstrated that Kcng4b modulates proliferation of cells lining the ventricular system and maintains their integrity. The gain of Kcng4b function reduces the size of brain ventricles. Electrophysiological studies suggest that Kcng4b mediates its effects via an antagonistic interaction with Kcnb1, the homologue of the electrically active delayed rectifier potassium channel subunit Kv2.1. Mutation of kcnb1 reduces the size of the ventricular system and its gain of function causes hydrocephalus, which is opposite to the function of Kcng4b. This demonstrates the dynamic interplay between potassium channel subunits in the neuroepithelium as a novel and crucial regulator of ventricular development in the vertebrate brain.
Collapse
Affiliation(s)
| | - Elke Bocksteins
- Department for Biomedical Sciences, University of Antwerp, Wilrijk B-2610, Belgium
| | | | - Dirk Snyders
- Department for Biomedical Sciences, University of Antwerp, Wilrijk B-2610, Belgium
| | - Vladimir Korzh
- Institute of Molecular and Cell Biology, Singapore .,Department of Biological Sciences, National University of Singapore, 117543, Singapore
| |
Collapse
|
23
|
Bocksteins E. Kv5, Kv6, Kv8, and Kv9 subunits: No simple silent bystanders. J Gen Physiol 2016; 147:105-25. [PMID: 26755771 PMCID: PMC4727947 DOI: 10.1085/jgp.201511507] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 12/11/2015] [Indexed: 12/19/2022] Open
Abstract
Members of the electrically silent voltage-gated K(+) (Kv) subfamilies (Kv5, Kv6, Kv8, and Kv9, collectively identified as electrically silent voltage-gated K(+) channel [KvS] subunits) do not form functional homotetrameric channels but assemble with Kv2 subunits into heterotetrameric Kv2/KvS channels with unique biophysical properties. Unlike the ubiquitously expressed Kv2 subunits, KvS subunits show a more restricted expression. This raises the possibility that Kv2/KvS heterotetramers have tissue-specific functions, making them potential targets for the development of novel therapeutic strategies. Here, I provide an overview of the expression of KvS subunits in different tissues and discuss their proposed role in various physiological and pathophysiological processes. This overview demonstrates the importance of KvS subunits and Kv2/KvS heterotetramers in vivo and the importance of considering KvS subunits and Kv2/KvS heterotetramers in the development of novel treatments.
Collapse
Affiliation(s)
- Elke Bocksteins
- Laboratory for Molecular Biophysics, Physiology, and Pharmacology, Department for Biomedical Sciences, University of Antwerp, 2610 Antwerp, Belgium
| |
Collapse
|
24
|
Cobb MM, Austin DC, Sack JT, Trimmer JS. Cell Cycle-dependent Changes in Localization and Phosphorylation of the Plasma Membrane Kv2.1 K+ Channel Impact Endoplasmic Reticulum Membrane Contact Sites in COS-1 Cells. J Biol Chem 2015; 290:29189-201. [PMID: 26442584 DOI: 10.1074/jbc.m115.690198] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Indexed: 12/22/2022] Open
Abstract
The plasma membrane (PM) comprises distinct subcellular domains with diverse functions that need to be dynamically coordinated with intracellular events, one of the most impactful being mitosis. The Kv2.1 voltage-gated potassium channel is conditionally localized to large PM clusters that represent specialized PM:endoplasmic reticulum membrane contact sites (PM:ER MCS), and overexpression of Kv2.1 induces more exuberant PM:ER MCS in neurons and in certain heterologous cell types. Localization of Kv2.1 at these contact sites is dynamically regulated by changes in phosphorylation at one or more sites located on its large cytoplasmic C terminus. Here, we show that Kv2.1 expressed in COS-1 cells undergoes dramatic cell cycle-dependent changes in its PM localization, having diffuse localization in interphase cells, and robust clustering during M phase. The mitosis-specific clusters of Kv2.1 are localized to PM:ER MCS, and M phase clustering of Kv2.1 induces more extensive PM:ER MCS. These cell cycle-dependent changes in Kv2.1 localization and the induction of PM:ER MCS are accompanied by increased mitotic Kv2.1 phosphorylation at several C-terminal phosphorylation sites. Phosphorylation of exogenously expressed Kv2.1 is significantly increased upon metaphase arrest in COS-1 and CHO cells, and in a pancreatic β cell line that express endogenous Kv2.1. The M phase clustering of Kv2.1 at PM:ER MCS in COS-1 cells requires the same C-terminal targeting motif needed for conditional Kv2.1 clustering in neurons. The cell cycle-dependent changes in localization and phosphorylation of Kv2.1 were not accompanied by changes in the electrophysiological properties of Kv2.1 expressed in CHO cells. Together, these results provide novel insights into the cell cycle-dependent changes in PM protein localization and phosphorylation.
Collapse
Affiliation(s)
- Melanie M Cobb
- From the Departments of Neurobiology, Physiology, and Behavior
| | | | - Jon T Sack
- Physiology and Membrane Biology, and Anesthesiology and Pain Medicine, University of California Davis School of Medicine, Davis, California 95616
| | - James S Trimmer
- From the Departments of Neurobiology, Physiology, and Behavior, Physiology and Membrane Biology, and
| |
Collapse
|
25
|
Effects of BKCa and Kir2.1 Channels on Cell Cycling Progression and Migration in Human Cardiac c-kit+ Progenitor Cells. PLoS One 2015; 10:e0138581. [PMID: 26390131 PMCID: PMC4577111 DOI: 10.1371/journal.pone.0138581] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Accepted: 09/01/2015] [Indexed: 12/24/2022] Open
Abstract
Our previous study demonstrated that a large-conductance Ca2+-activated K+ current (BKCa), a voltage-gated TTX-sensitive sodium current (INa.TTX), and an inward rectifier K+ current (IKir) were heterogeneously present in most of human cardiac c-kit+ progenitor cells. The present study was designed to investigate the effects of these ion channels on cell cycling progression and migration of human cardiac c-kit+ progenitor cells with approaches of cell proliferation and mobility assays, siRNA, RT-PCR, Western blots, flow cytometry analysis, etc. It was found that inhibition of BKCa with paxilline, but not INa.TTX with tetrodotoxin, decreased both cell proliferation and migration. Inhibition of IKir with Ba2+ had no effect on cell proliferation, while enhanced cell mobility. Silencing KCa.1.1 reduced cell proliferation by accumulating the cells at G0/G1 phase and decreased cell mobility. Interestingly, silencing Kir2.1 increased the cell migration without affecting cell cycling progression. These results demonstrate the novel information that blockade or silence of BKCa channels, but not INa.TTX channels, decreases cell cycling progression and mobility, whereas inhibition of Kir2.1 channels increases cell mobility without affecting cell cycling progression in human cardiac c-kit+ progenitor cells.
Collapse
|
26
|
Ferreira R, Wong R, Schlichter LC. KCa3.1/IK1 Channel Regulation by cGMP-Dependent Protein Kinase (PKG) via Reactive Oxygen Species and CaMKII in Microglia: An Immune Modulating Feedback System? Front Immunol 2015; 6:153. [PMID: 25904916 PMCID: PMC4389654 DOI: 10.3389/fimmu.2015.00153] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 03/23/2015] [Indexed: 01/09/2023] Open
Abstract
The intermediate conductance Ca2+-activated K+ channel, KCa3.1 (IK1/SK4/KCNN4) is widely expressed in the innate and adaptive immune system. KCa3.1 contributes to proliferation of activated T lymphocytes, and in CNS-resident microglia, it contributes to Ca2+ signaling, migration, and production of pro-inflammatory mediators (e.g., reactive oxygen species, ROS). KCa3.1 is under investigation as a therapeutic target for CNS disorders that involve microglial activation and T cells. However, KCa3.1 is post-translationally regulated, and this will determine when and how much it can contribute to cell functions. We previously found that KCa3.1 trafficking and gating require calmodulin (CaM) binding, and this is inhibited by cAMP kinase (PKA) acting at a single phosphorylation site. The same site is potentially phosphorylated by cGMP kinase (PKG), and in some cells, PKG can increase Ca2+, CaM activation, and ROS. Here, we addressed KCa3.1 regulation through PKG-dependent pathways in primary rat microglia and the MLS-9 microglia cell line, using perforated-patch recordings to preserve intracellular signaling. Elevating cGMP increased both the KCa3.1 current and intracellular ROS production, and both were prevented by the selective PKG inhibitor, KT5823. The cGMP/PKG-evoked increase in KCa3.1 current in intact MLS-9 microglia was mediated by ROS, mimicked by applying hydrogen peroxide (H2O2), inhibited by a ROS scavenger (MGP), and prevented by a selective CaMKII inhibitor (mAIP). Similar results were seen in alternative-activated primary rat microglia; their KCa3.1 current required PKG, ROS, and CaMKII, and they had increased ROS production that required KCa3.1 activity. The increase in current apparently did not result from direct effects on the channel open probability (Po) or Ca2+ dependence because, in inside-out patches from transfected HEK293 cells, single-channel activity was not affected by cGMP, PKG, H2O2 at normal or elevated intracellular Ca2+. The regulation pathway we have identified in intact microglia and MLS-9 cells is expected to have broad implications because KCa3.1 plays important roles in numerous cells and tissues.
Collapse
Affiliation(s)
- Roger Ferreira
- Genetics and Development Division, Toronto Western Research Institute, University Health Network , Toronto, ON , Canada ; Department of Physiology, University of Toronto , Toronto, ON , Canada
| | - Raymond Wong
- Genetics and Development Division, Toronto Western Research Institute, University Health Network , Toronto, ON , Canada ; Department of Physiology, University of Toronto , Toronto, ON , Canada
| | - Lyanne C Schlichter
- Genetics and Development Division, Toronto Western Research Institute, University Health Network , Toronto, ON , Canada ; Department of Physiology, University of Toronto , Toronto, ON , Canada
| |
Collapse
|
27
|
The role of KCa3.1 channels in cardiac fibrosis induced by pressure overload in rats. Pflugers Arch 2015; 467:2275-85. [DOI: 10.1007/s00424-015-1694-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 02/10/2015] [Accepted: 02/11/2015] [Indexed: 10/23/2022]
|
28
|
Prasad M, Zachar V, Fink T, Pennisi CP. Moderate hypoxia influences potassium outward currents in adipose-derived stem cells. PLoS One 2014; 9:e104912. [PMID: 25115627 PMCID: PMC4130641 DOI: 10.1371/journal.pone.0104912] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 07/15/2014] [Indexed: 01/23/2023] Open
Abstract
Moderate hypoxic preconditioning of adipose-derived stem cells (ASCs) enhances properties such as proliferation and secretion of growth factors, representing a valuable strategy to increase the efficiency of cell-based therapies. In a wide variety of cells potassium (K+) channels are key elements involved in the cellular responses to hypoxia, suggesting that ASCs cultured under low oxygen conditions may display altered electrophysiological properties. Here, the effects of moderate hypoxic culture on proliferation, whole-cell currents, and ion channel expression were investigated using human ASCs cultured at 5% and 20% oxygen. Although cell proliferation was greatly enhanced, the dose-dependent growth inhibition by the K+ channel blocker tetraethylammonium (TEA) was not significantly affected by hypoxia. Under both normoxic and hypoxic conditions, ASCs displayed outward K+ currents composed by Ca2+-activated, delayed rectifier, and transient components. Hypoxic culture reduced the slope of the current-voltage curves and caused a negative shift in the voltage activation threshold of the whole-cell currents. However, the TEA-mediated shift of voltage activation threshold was not affected by hypoxia. Semiquantitative real-time RT-PCR revealed that expression of genes encoding for various ion channels subunits related to oxygen sensing and proliferation remained unchanged after hypoxic culture. In conclusion, outward currents are influenced by moderate hypoxia in ASCs through a mechanism that is not likely the result of modulation of TEA-sensitive K+ channels.
Collapse
Affiliation(s)
- Mayuri Prasad
- Laboratory for Stem Cell Research, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Vladimir Zachar
- Laboratory for Stem Cell Research, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Trine Fink
- Laboratory for Stem Cell Research, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Cristian Pablo Pennisi
- Laboratory for Stem Cell Research, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
- * E-mail:
| |
Collapse
|
29
|
Che H, Yue J, Tse HF, Li GR. Functional TRPV and TRPM channels in human preadipocytes. Pflugers Arch 2014; 466:947-959. [PMID: 24057349 DOI: 10.1007/s00424-013-1355-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 09/09/2013] [Accepted: 09/09/2013] [Indexed: 12/30/2022]
Abstract
Preadipocytes are widely used as an in vitro model to investigate proliferation, adipogenic differentiation, and lipodystrophy; however, cellular physiology and biology are not fully understood in human preadipocytes. The present study was to investigate the expression of transient receptor potential (TRP) channels in human preadipocytes and their potential roles in regulating proliferation and adipogenic differentiation using approaches of confocal microscopy, whole-cell patch voltage-clamp, reverse transcription polymerase chain reaction, Western blot, etc. We found that TRPV2, TRPV4, and TRPM7 channels were abundantly expressed in human preadipocytes. The intracellular Ca(2+) transient activated by the TRPV2 activator probenecid was reversed or prevented by ruthenium red, a TRPV2 blocker. The TRPV4 channel activator, 4α-phorbol 12-13-dicaprinate, enhanced intracellular Ca(2+) oscillations, and the effect was inhibited by the TRPV4 blocker RN-1734. TRPM7 current was recorded with dialysis of Mg(2+)-free pipette solution, which was inhibited by the TRP channel blocker 2-aminoethoxydiphenyl borate and enhanced by acidic extracellular pH. Silencing TRPV2 or TRPM7, but not TRPV4, significantly reduced cell proliferation via inhibiting cyclin D1, cyclin E, and p-ERK1/2. Interestingly, individually silencing these three channels decreased adipogenic differentiation of human preadipocytes by reducing p-Akt kinase. Our results demonstrate for the first time that functional TRPV2, TRPV4, and TRPM7 channels are abundantly expressed in human preadipocytes. TRPV2 and TRPM7, but not TRPV4, regulate cell proliferation via activating cyclin D1, cyclin E, and p-ERK1/2, while they are all involved in adipogenesis in human preadipocytes via phosphorylating Akt kinase.
Collapse
Affiliation(s)
- Hui Che
- Department of Medicine, Li-Ka Shing Faculty of Medicine, University of Hong Kong, Laboratory Block, FMB, 21 Sassoon Road, Pokfulam, Hong Kong, China
| | | | | | | |
Collapse
|
30
|
Zhang YY, Li G, Che H, Sun HY, Li X, Au WK, Xiao GS, Wang Y, Li GR. Characterization of functional ion channels in human cardiac c-kit+ progenitor cells. Basic Res Cardiol 2014; 109:407. [DOI: 10.1007/s00395-014-0407-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 01/27/2014] [Accepted: 02/25/2014] [Indexed: 12/17/2022]
|
31
|
Zhang YY, Yue J, Che H, Sun HY, Tse HF, Li GR. BKCa and hEag1 channels regulate cell proliferation and differentiation in human bone marrow-derived mesenchymal stem cells. J Cell Physiol 2014; 229:202-212. [PMID: 23881642 DOI: 10.1002/jcp.24435] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 07/15/2013] [Indexed: 12/29/2022]
Abstract
Human bone marrow-derived mesenchymal stem cells (MSCs) serve as a reservoir for the continuous renewal of various mesenchymal tissues; however, cellular physiology of ion channels is not fully understood. The present study investigated potential roles of large-conductance Ca(2+) -activated potassium (BKCa ) channels and ether-à-go-go potassium (hEag1 or Kv10.1) channels in regulating cell proliferation and differentiation in human MSCs. We found that inhibition of BKCa with paxilline or hEag1 with astemizole, or knockdown of BKCa with shRNAs targeting KCa1.1 or hEag1 channels with shRNAs targeting KCNH1 arrested the cells at G0/G1 phase. In addition, silencing BKCa or hEag1 channels significantly reduced adipogenic differentiation with decrease of lipid accumulation and expression of the adipocyte marker PPARγ, and decreased osteogenic differentiation with reduction of mineral precipitation and osteocalcin. These effects were accompanied with a reduced cyclin D1, cyclin E, p-ERK1/2, and p-Akt. Our results demonstrate that BKCa and hEag1 channels not only regulate cell proliferation, but also participate in the adipogenic and osteogenic differentiations in human MSCs, which indicates that BKCa and hEag1 channels may be essential in maintaining bone marrow physiological function and bone regeneration.
Collapse
Affiliation(s)
- Ying-Ying Zhang
- Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | | | | | | | | | | |
Collapse
|
32
|
Up-regulation of KCa3.1 promotes human airway smooth muscle cell phenotypic modulation. Pharmacol Res 2013; 77:30-8. [PMID: 24055799 DOI: 10.1016/j.phrs.2013.09.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 08/26/2013] [Accepted: 09/08/2013] [Indexed: 02/06/2023]
Abstract
Airway smooth muscle (ASM) cell phenotype modulation, characterized by reversible switching between contractile and proliferative phenotypes, is considered to contribute to proliferative diseases such as allergic asthma and chronic obstructive pulmonary disease (COPD). KCa3.1 has been suggested to be involved in regulating ASM cell activation, proliferation, and migration. However, little is known regarding the exact role of KCa3.1 in ASM cell phenotypic modulation. To elucidate the role of KCa3.1 in regulating ASM cell phenotypic modulation, we investigated the effects of KCa3.1 channels on ASM contractile marker protein expression, proliferation and migration of primary human bronchial smooth muscle (BSM) cells. We found that PDGF increased KCa3.1 channel expression in BSM cells with a concomitant marked decrease in the expression of contractile phenotypic marker proteins including smooth muscle myosin heavy chain (SMMHC), smooth muscle α-actin (α-SMA), myocardin and KCa1.1. These changes were significantly attenuated by the KCa3.1 blocker, TRAM-34, or gene silencing of KCa3.1. Pharmacological blockade or gene silencing of KCa3.1 also suppressed PDGF-induced human BSM cell migration and proliferation accompanied by a decrease in intracellular free Ca(2+) levels as a consequence of membrane depolarization, resulting in a reduction in cyclin D1 level and cell cycle arrest at G0-G1 phase. Additionally, PDGF-induced up-regulation of KCa3.1 and down-regulation of BSM contractile marker proteins were regulated by the ERK inhibitor U0126 and the AKT inhibitor LY294002. These findings highlight a novel role for the KCa3.1 channel in human BSM cell phenotypic modulation and provide a potential target for therapeutic intervention for proliferative airway diseases.
Collapse
|
33
|
Wang L, Yan M, Wang Y, Lei G, Yu Y, Zhao C, Tang Z, Zhang G, Tang C, Yu J, Liao H. Proliferation and osteo/odontoblastic differentiation of stem cells from dental apical papilla in mineralization-inducing medium containing additional KH(2)PO(4). Cell Prolif 2013; 46:214-22. [PMID: 23510476 DOI: 10.1111/cpr.12016] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 11/16/2012] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES Stem cells from the dental apical papilla (SCAPs) can be induced to differentiate along both osteoblast and odontoblast lineages. However, little knowledge is available concerning their differentiation efficiency in osteogenic media containing additional KH2 PO4 . MATERIALS AND METHODS Stem cells from the dental apical papilla were isolated from apical papillae of immature third molars and treated with two kinds of mineralization-inducing media, MM1 and MM2, differing in KH2 PO4 concentration. Proliferation and osteo/odontogenic differentiation capacity of MM1/MM2-treated SCAPs were investigated and compared both in vitro and in vivo. RESULTS Cell counting and flow cytometry demonstrated that MM2 containing 1.8 mm additional KH2 PO4 significantly enhanced proliferative potential of SCAPs, compared to MM1. Osteo/odontogenic capacity of SCAPs was much better in MM2 medium than in MM1, as indicated by elevated alkaline phosphatase activity, increased calcium deposition and upregulated expression of osteo/odontoblast-specific genes/proteins (for example, runt-related transcription factor 2, osterix, osteocalcin, dentin sialoprotein, and dentin sialophosphoprotein). In vivo transplantation findings proved that SCAPs in MM2 group generated more mineralized tissues, and presented higher expression of osteo/odontoblast-specific proteins (osteocalcin and dentin sialoprotein) than those in the MM1 group. CONCLUSION Mineralization-inducing media supplemented with 1.8 mm additional KH2 PO4 significantly enhanced cell proliferation and improved differentiation capacity of SCAPs along osteo/odontogenic cell lineages, compared to counterparts lacking additional KH2 PO4 .
Collapse
Affiliation(s)
- L Wang
- Institute of Stomatology, Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Wang LP, Wang Y, Zhao LM, Li GR, Deng XL. Angiotensin II upregulates K(Ca)3.1 channels and stimulates cell proliferation in rat cardiac fibroblasts. Biochem Pharmacol 2013; 85:1486-94. [PMID: 23500546 DOI: 10.1016/j.bcp.2013.02.032] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 02/20/2013] [Accepted: 02/27/2013] [Indexed: 12/01/2022]
Abstract
The proliferation of cardiac fibroblasts is implicated in the pathogenesis of myocardial remodeling and fibrosis. Intermediate-conductance calcium-activated K⁺ channels (K(Ca)3.1 channels) have important roles in cell proliferation. However, it is unknown whether angiotensin II (Ang II), a potent profibrotic molecule, would regulate K(Ca)3.1 channels in cardiac fibroblasts and participate in cell proliferation. In the present study, we investigated whether K(Ca)3.1 channels were regulated by Ang II, and how the channel activity mediated cell proliferation in cultured adult rat cardiac fibroblasts using electrophysiology and biochemical approaches. It was found that mRNA, protein, and current density of K(Ca)3.1 channels were greatly enhanced in cultured cardiac fibroblasts treated with 1 μM Ang II, and the effects were countered by the angiotensin type 1 receptor (AT₁R) blocker losartan, the p38-MAPK inhibitor SB203580, the ERK1/2 inhibitor PD98059, and the PI3K/Akt inhibitor LY294002. Ang II stimulated cell proliferation and the effect was antagonized by the K(Ca)3.1 blocker TRAM-34 and siRNA targeting K(Ca)3.1. In addition, Ang II-induced increase of K(Ca)3.1 expression was attenuated by transfection of activator protein-1 (AP-1) decoy oligodeoxynucleotides. These results demonstrate for the first time that Ang II stimulates cell proliferation mediated by upregulating K(Ca)3.1 channels via interacting with the AT₁R and activating AP-1 complex through ERK1/2, p38-MAPK and PI3K/Akt signaling pathways in cultured adult rat cardiac fibroblasts.
Collapse
Affiliation(s)
- Li-Ping Wang
- Department of Physiology and Pathophysiology, School of Medicine, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, 710061 Shaanxi, China
| | | | | | | | | |
Collapse
|
35
|
Zhao LM, Su XL, Wang Y, Li GR, Deng XL. KCa3.1 channels mediate the increase of cell migration and proliferation by advanced glycation endproducts in cultured rat vascular smooth muscle cells. J Transl Med 2013; 93:159-67. [PMID: 23212096 DOI: 10.1038/labinvest.2012.163] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The mechanisms underlying the involvement of advanced glycation endproducts (AGEs) in diabetic atherosclerosis are not fully understood. The present study was designed to investigate whether intermediate-conductance Ca(2+)-activated K(+) channels (K(Ca)3.1 channels) are involved in migration and proliferation induced by AGEs in cultured rat vascular smooth muscle cells (VSMCs) using approaches of whole-cell patch voltage-clamp, cell proliferation and migration assay, and western blot analysis. It was found that the current density and protein level of K(Ca)3.1 channels were enhanced in cells incubated with AGE-BSA (bovine serum albumin), and the effects were reversed by co-incubation of AGE-BSA with anti-RAGE (anti-receptors of AGEs) antibody. The ERK1/2 inhibitors PD98059 and U0126, the P38-MAPK inhibitors SB203580 and SB202190, or the PI3K inhibitors LY294002 and wortmannin countered the K(Ca)3.1 channel expression by AGE-BSA. In addition, AGE-BAS increased cell migration and proliferation, and the effects were fully reversed with anti-RAGE antibody, the K(Ca)3.1 channel blocker TRAM-34, or K(Ca)3.1 small interfering RNA. These results demonstrate for the first time that AGEs-induced increase of migration and proliferation is related to the upregulation of K(Ca)3.1 channels in rat VMSCs, and the intracellular signals ERK1/2, P38-MAPK and PI3K are involved in the regulation of K(Ca)3.1 channel expression.
Collapse
Affiliation(s)
- Li-Mei Zhao
- Department of Physiology and Pathophysiology, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | | | | | | | | |
Collapse
|
36
|
Chen JB, Liu WJ, Che H, Liu J, Sun HY, Li GR. Adenosine-5'-triphosphate up-regulates proliferation of human cardiac fibroblasts. Br J Pharmacol 2012; 166:1140-50. [PMID: 22224416 DOI: 10.1111/j.1476-5381.2012.01831.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND AND PURPOSE ATP is a potent signalling molecule that regulates biological activities including increasing or decreasing proliferation in different types of cells. The aim of the present study was to investigate how ATP regulates the proliferation of human cardiac fibroblasts. EXPERIMENTAL APPROACH Reverse transcription (RT)-PCR, Western blot analysis, cell proliferation and migration assays were employed to investigate the effects of ATP on human adult ventricular fibroblasts. KEY RESULTS ATP increased cell proliferation in a concentration-dependent manner. Similarly, the P2X receptor agonist α,β-methylene ATP and P2Y receptor agonist ATP-γS also up-regulated cell proliferation. The P2 receptor antagonists suramin and reactive blue-2 prevented the ATP-induced increase in proliferation and RT-PCR and Western blot analysis revealed that mRNAs of P2X(4/7) and P2Y(2) are abundant in cardiac fibroblasts. ATP increased phosphorylated PKB (Akt) and ERK1/2 levels; an effect antagonized by suramin, reactive blue-2, the PI3-kinase inhibitor, wortmannin, PKB inhibitor, API-2, and MAPK inhibitor, PD98059. These kinase inhibitors also prevented the ATP-induced increase in proliferation. In addition, ATP enhanced the progression of cells from the G0/G1 phase to the S phase by increasing the expression of proteins for cyclin D1 and cyclin E. Silencing the P2X(4/7) and P2Y(2) receptors with siRNA targeting the corresponding receptor diminished ATP-stimulated proliferation and migration of the cardiac fibroblasts. CONCLUSION AND IMPLICATION ATP activates P2X(4/7) and P2Y(2) receptors and up-regulates the proliferation of human cardiac fibroblasts by promoting cell cycling progression. It also increases the migration of these cells. These effects of ATP may be involved in cardiac remodelling of injured hearts.
Collapse
Affiliation(s)
- Jing-Bo Chen
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | | | | | | | | | | |
Collapse
|
37
|
Zhao LM, Zhang W, Wang LP, Li GR, Deng XL. Advanced glycation end products promote proliferation of cardiac fibroblasts by upregulation of KCa3.1 channels. Pflugers Arch 2012; 464:613-21. [PMID: 23053478 DOI: 10.1007/s00424-012-1165-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2012] [Revised: 09/17/2012] [Accepted: 09/19/2012] [Indexed: 01/02/2023]
Abstract
The present study was designed to investigate whether advanced glycation end products (AGEs) would regulate K(Ca)3.1 channels in cardiac fibroblasts and participate in cell proliferation. Cultured adult rat cardiac fibroblasts were employed to investigate the regulation of K(Ca)3.1 channels by advanced glycation end products-bovine serum albumin (AGE-BSA) and the role of K(Ca)3.1 channels in cell proliferation using approaches of molecular biology. K(Ca)3.1 channel mRNA and protein levels were greatly enhanced in cardiac fibroblasts treated with 200 μg/ml AGE-BSA, and the effects were countered by anti-RAGE antibody or the ERK1/2 inhibitor PD98059, the p38-MAPK inhibitor SB203580, and the PI3K/Akt inhibitor LY294002. In addition, AGE-BSA stimulated cell proliferation and collagen production in cultured cardiac fibroblasts, and the effects were reversed by K(Ca)3.1 blocker TRAM-34, anti-RAGE antibody, or signal inhibitors PD98059, SB203580, and LY294002. These results demonstrate for the first time that AGEs increase the expression of K(Ca)3.1 channels in a RAGE-dependent manner and promote cardiac fibroblast proliferation and collagen production, which is mediated by phosphorylation of ERK1/2, p38-MAPK, and PI3K/Akt signals.
Collapse
Affiliation(s)
- Li-Mei Zhao
- Department of Physiology and Pathophysiology, School of Medicine, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, Shaanxi, China
| | | | | | | | | |
Collapse
|
38
|
Ion channels in hematopoietic and mesenchymal stem cells. Stem Cells Int 2012; 2012:217910. [PMID: 22919401 PMCID: PMC3420091 DOI: 10.1155/2012/217910] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Accepted: 07/05/2012] [Indexed: 12/19/2022] Open
Abstract
Hematopoietic stem cells (HSCs) reside in bone marrow niches and give rise to hematopoietic precursor cells (HPCs). These have more restricted lineage potential and eventually differentiate into specific blood cell types. Bone marrow also contains mesenchymal stromal cells (MSCs), which present multilineage differentiation potential toward mesodermal cell types. In bone marrow niches, stem cell interaction with the extracellular matrix is mediated by integrin receptors. Ion channels regulate cell proliferation and differentiation by controlling intracellular Ca(2+), cell volume, release of growth factors, and so forth. Although little evidence is available about the ion channel roles in true HSCs, increasing information is available about HPCs and MSCs, which present a complex pattern of K(+) channel expression. K(+) channels cooperate with Ca(2+) and Cl(-) channels in regulating calcium entry and cell volume during mitosis. Other K(+) channels modulate the integrin-dependent interaction between leukemic progenitor cells and the niche stroma. These channels can also regulate leukemia cell interaction with MSCs, which also involves integrin receptors and affects the MSC-mediated protection from chemotherapy. Ligand-gated channels are also implicated in these processes. Nicotinic acetylcholine receptors regulate cell proliferation and migration in HSCs and MSCs and may be implicated in the harmful effects of smoking.
Collapse
|
39
|
Li X, Zhang M, Bai L, Bai W, Xu W, Zhu H. Effects of 50 Hz pulsed electromagnetic fields on the growth and cell cycle arrest of mesenchymal stem cells: an in vitro study. Electromagn Biol Med 2012; 31:356-64. [PMID: 22676915 DOI: 10.3109/15368378.2012.662194] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Xinping Li
- Department of Rehabilitation, Guangdong Geriatric Institute, Guangdong Academy of Medical Sciences and Guangdong General Hospital
Guangzhou, China
| | - Mingsheng Zhang
- Department of Rehabilitation, Guangdong Geriatric Institute, Guangdong Academy of Medical Sciences and Guangdong General Hospital
Guangzhou, China
| | - Liming Bai
- Department of Rehabilitation, Guangdong Geriatric Institute, Guangdong Academy of Medical Sciences and Guangdong General Hospital
Guangzhou, China
| | - Wenfang Bai
- Department of Rehabilitation, Guangdong Geriatric Institute, Guangdong Academy of Medical Sciences and Guangdong General Hospital
Guangzhou, China
| | - Weicheng Xu
- Department of Rehabilitation, Guangdong Geriatric Institute, Guangdong Academy of Medical Sciences and Guangdong General Hospital
Guangzhou, China
| | - Hongxiang Zhu
- Department of Rehabilitation, Guangdong Geriatric Institute, Guangdong Academy of Medical Sciences and Guangdong General Hospital
Guangzhou, China
| |
Collapse
|
40
|
Xu X, Xia J, Yang X, Huang X, Gao D, Zhou J, Lian J, Zhou J. Intermediate-conductance Ca(2+) -activated potassium and volume-sensitive chloride channels in endothelial progenitor cells from rat bone marrow mononuclear cells. Acta Physiol (Oxf) 2012; 205:302-13. [PMID: 22168445 DOI: 10.1111/j.1748-1716.2011.02398.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Revised: 02/17/2011] [Accepted: 12/08/2011] [Indexed: 12/24/2022]
Abstract
AIM Bone marrow endothelial progenitor cells (BMEPCs) are believed to be a promising cell source for regenerative medicine; however, their electrophysiology properties have not been fully clarified, which is important to the clinical application of BMEPCs. The current study was designed to determine the transmembrane ion currents and mRNA expression levels of related ion channel subunits in rat BMEPCs. METHODS Bone marrow mononuclear cells were isolated by density gradient separation and cultured in EPC medium. The transmembrane ion currents were determined using whole-cell patch-voltage clamp technique, and the levels of mRNA and protein expressions of functional ionic channels were measured using RT-PCR and western immunoblot analysis. RESULTS We observed two types of ionic currents in undifferentiated rat BMEPCs. One was Ca(2+) -activated potassium current (I(kca) ), which was seen in approx. 90% of cells when 1 μm Ca(2+) was employed in pipette solution, and it was predominantly inhibited by intermediate-conductance I(kca) inhibitor clotrimazole. The other one was volume-sensitive chloride current (I(cl) ), which was detected in 85.7% of cells when BMEPCs were subjected to K(+) -free hypotonic extracellular solution, whose currents could be inhibited by 5-nitro-2-(3-phenylpropylamino) benzoic acid (NPPB). The corresponding ion channel genes and proteins, KCNN4 for I(kca) and Clcn3 for I(cl) , were confirmed by RT-PCR and western immunoblot analysis of BMEPCs. CONCLUSION Our results demonstrated for the first time that rat BMEPCs expressed intermediate-conductance Ca(2+) -activated potassium currents and volume-sensitive chloride currents, and corresponding genes and proteins of these two channels are KCNN4 and Clcn3 respectively.
Collapse
Affiliation(s)
- X. Xu
- Lihuili Hospital; Ningbo University School of Medicine; Ningbo; China
| | - J. Xia
- Lihuili Hospital; Ningbo University School of Medicine; Ningbo; China
| | - X. Yang
- Lihuili Hospital; Ningbo University School of Medicine; Ningbo; China
| | - X. Huang
- Lihuili Hospital; Ningbo University School of Medicine; Ningbo; China
| | - D. Gao
- Lihuili Hospital; Ningbo University School of Medicine; Ningbo; China
| | - J. Zhou
- Lihuili Hospital; Ningbo University School of Medicine; Ningbo; China
| | - J. Lian
- Lihuili Hospital; Ningbo University School of Medicine; Ningbo; China
| | - J. Zhou
- Lihuili Hospital; Ningbo University School of Medicine; Ningbo; China
| |
Collapse
|
41
|
Zhang XH, Zhang YY, Sun HY, Jin MW, Li GR. Functional ion channels and cell proliferation in 3T3-L1 preadipocytes. J Cell Physiol 2012; 227:1972-9. [DOI: 10.1002/jcp.22925] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
42
|
Liebau S, Tischendorf M, Ansorge D, Linta L, Stockmann M, Weidgang C, Iacovino M, Boeckers T, von Wichert G, Kyba M, Kleger A. An inducible expression system of the calcium-activated potassium channel 4 to study the differential impact on embryonic stem cells. Stem Cells Int 2011; 2011:456815. [PMID: 21941566 PMCID: PMC3173888 DOI: 10.4061/2011/456815] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Accepted: 06/14/2011] [Indexed: 11/20/2022] Open
Abstract
Rationale. The family of calcium-activated potassium channels consists of four members with varying biological functions and conductances. Besides membrane potential modulation, SK channels have been found to be involved in cardiac pacemaker cell development from ES cells and morphological shaping of neural stem cells. Objective. Distinct SK channel subtype expression in ES cells might elucidate their precise impact during cardiac development. We chose SK channel subtype 4 as a potential candidate influencing embryonic stem cell differentiation. Methods. We generated a doxycycline inducible mouse ES cell line via targeted homologous recombination of a cassette expressing a bicistronic construct encoding SK4 and a fluorophore from the murine HPRT locus. Conclusion. We characterized the mouse ES cell line iSK4-AcGFP. The cassette is readily expressed under the control of doxycycline, and the overexpression of SK4 led to an increase in cardiac and pacemaker cell differentiation thereby serving as a unique tool to characterize the cell biological variances due to specific SK channel overexpression.
Collapse
Affiliation(s)
- Stefan Liebau
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Effects of ion channels on proliferation in cultured human cardiac fibroblasts. J Mol Cell Cardiol 2011; 51:198-206. [DOI: 10.1016/j.yjmcc.2011.05.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Revised: 04/24/2011] [Accepted: 05/11/2011] [Indexed: 01/01/2023]
|
44
|
Insulin-mediated upregulation of KCa3.1 channels promotes cell migration and proliferation in rat vascular smooth muscle. J Mol Cell Cardiol 2011; 51:51-7. [DOI: 10.1016/j.yjmcc.2011.03.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Revised: 03/07/2011] [Accepted: 03/26/2011] [Indexed: 12/29/2022]
|
45
|
Hu X, Wei L, Taylor TM, Wei J, Zhou X, Wang JA, Yu SP. Hypoxic preconditioning enhances bone marrow mesenchymal stem cell migration via Kv2.1 channel and FAK activation. Am J Physiol Cell Physiol 2011; 301:C362-72. [PMID: 21562308 DOI: 10.1152/ajpcell.00013.2010] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Transplantation using stem cells including bone marrow mesenchymal stem cells (BMSCs) is emerging as a potential regenerative therapy after ischemic attacks in the heart and brain. The migration capability of transplanted cells is a critical cellular function for tissue repair. Based on our recent observations that hypoxic preconditioning (HP) has multiple benefits in improving stem cell therapy and that the potassium Kv2.1 channel acts as a promoter for focal adhesion kinase (FAK) activation and cell motility, the present investigation tested the hypothesis that HP treatment can increase BMSC migration via the mechanism of increased Kv2.1 expression and FAK activities. BMSCs derived from green fluorescent protein-transgenic mice were treated under either normoxic (N-BMSC) or hypoxic (0.5% O(2)) (HP-BMSC) conditions for 24 h. Western blot analysis showed HP selectively upregulated Kv2.1 expression while leaving other K(+) channels, such as Kv1.5 and Kv1.4, unaffected. Compared with normoxic controls, significantly larger outward delayed rectifier K(+) currents were recorded in HP-BMSCs. HP enhanced BMSC migration/homing activities in vitro and after intravenous transplantation into rats subjected to permanent myocardial infarction (MI). The HP-promoted BMSC migration was inhibited by either blocking K(+) channels or knocking down Kv2.1. Supporting a relationship among HP, Kv2.1, and FAK activation, HP increased phosphorylation of FAK(397) and FAK(576/577), and this effect was antagonized by blocking K(+) channels. These findings provide novel evidence that HP enhances the ability of BMSCs to migrate and home to the injured region; this effect is mediated through a regulatory role of Kv2.1 on FAK phosphorylation/activation.
Collapse
Affiliation(s)
- Xinyang Hu
- Department of Cardiology, Second Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, China
| | | | | | | | | | | | | |
Collapse
|
46
|
Li GR, Deng XL. Functional ion channels in stem cells. World J Stem Cells 2011; 3:19-24. [PMID: 21607133 PMCID: PMC3097936 DOI: 10.4252/wjsc.v3.i3.19] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2010] [Revised: 01/14/2011] [Accepted: 01/21/2011] [Indexed: 02/06/2023] Open
Abstract
Bioelectrical signals generated by ion channels play crucial roles in excitation genesis and impulse conduction in excitable cells as well as in cell proliferation, migration and apoptosis in proliferative cells. Recent studies have demonstrated that multiple ion channels are heterogeneously present in different stem cells; however, patterns and phenotypes of ion channels are species- and/or origin-dependent. This editorial review focuses on the recent findings related to the expression of functional ion channels and the roles of these channels in regulation of cell proliferation in stem cells. Additional effort is required in the future to clarify the ion channel expression in different types of stem cells; special attention should be paid to the relationship between ion channels and stem cell proliferation, migration and differentiation.
Collapse
Affiliation(s)
- Gui-Rong Li
- Gui-Rong Li, Departments of Medicine and Physiology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong, China
| | | |
Collapse
|
47
|
Erythropoietin attenuates inflammatory factors and cell death in neonatal rats with intracerebral hemorrhage. ACTA NEUROCHIRURGICA. SUPPLEMENT 2011; 111:299-305. [PMID: 21725772 DOI: 10.1007/978-3-7091-0693-8_50] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Stroke affects infants at a rate of 26/100,000 live births each year. Of these strokes, approximately 6.7 are hemorrhagic strokes. Erythropoietin (EPO) is an anti-apoptotic and neuroprotective hormone. In adult rodents, EPO attenuates inflammatory factor expression and blood-brain barrier damage after intracerebral hemorrhage (ICH). However, the effect of EPO in neonatal ICH stroke remains unexplored. This investigation aimed to elucidate the underpinnings of inflammation after ICH in postnatal day 7 (P7) rats and the effect of human recombinant EPO (hrEPO) treatment on ICH-induced inflammation. The P7 rat pups were pretreated with hrEPO (5,000 U/kg i.p.) or saline vehicle 4 h prior to the induction of ICH by blood injection into the right cerebral cortex and basal ganglia. Supplemental half doses of hrEPO treatment or saline injections were subsequently given 16 h after ICH induction. Real-time PCR done 24 h after ICH showed reductions in interleukin1-β (IL1-β), interleukin-6 (IL-6) and tumor necrosis factor-α (TNFα) mRNA expression in the basal ganglia of the hrEPO-treated rats compared to saline-treated rats. Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining indicated fewer dying cells in the hrEPO-treated brain. Our data suggest that hrEPO has an anti-inflammatory action in neonates after ICH. The suppression of inflammatory cascades likely contributes to hrEPO's neuroprotective effect, which may be explored as a therapeutic treatment for ICH.
Collapse
|
48
|
Choi JH, Lim CJ, Han TH, Lee SK, Lee SY, Ryu PD. TEA-sensitive currents contribute to membrane potential of organ surface primo-node cells in rats. J Membr Biol 2010; 239:167-75. [PMID: 21153632 DOI: 10.1007/s00232-010-9335-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Accepted: 11/16/2010] [Indexed: 11/29/2022]
Abstract
The primo-vascular (Bonghan) tissue has been identified in most tissues in the body, but its structure and functions are not yet well understood. We characterized electrophysiological properties of the cells of the primo-nodes (PN) on the surface of abdominal organs using a slice patch clamp technique. The most abundant were small round cells (~10 μm) without processes. These PN cells exhibited low resting membrane potential (-36 mV) and did not fire action potentials. On the basis of the current-voltage (I-V) relationships and kinetics of outward currents, the PN cells can be grouped into four types. Among these, type I cells were the majority (69%); they showed strong outward rectification in I-V relations. The outward current was activated rapidly and sustained without decay. Tetraethylammonium (TEA) dose-dependently blocked both outward and inward current (IC(50), 4.3 mM at ± 60 mV). In current clamp conditions, TEA dose-dependently depolarized the membrane potential (18.5 mV at 30 mM) with increase in input resistance. The tail current following a depolarizing voltage step was reversed at -27 mV, and transient outward current like A-type K(+) current was not expressed at holding potential of -80 mV. Taken together, the results demonstrate for the first time that the small round PN cells are heterogenous, and that, in type I cells, TEA-sensitive current with limited selectivity to K(+) contributed to resting membrane potential of these cells.
Collapse
Affiliation(s)
- Jae-Hong Choi
- Laboratory of Veterinary Pharmacology, Research Institute of Veterinary Science, College of Veterinary Medicine, Seoul National University, 599 Gwanak-Ro, Gwanak-Gu, Seoul 151-742, Republic of Korea
| | | | | | | | | | | |
Collapse
|
49
|
Sun LY, Hsieh DK, Lin PC, Chiu HT, Chiou TW. Pulsed electromagnetic fields accelerate proliferation and osteogenic gene expression in human bone marrow mesenchymal stem cells during osteogenic differentiation. Bioelectromagnetics 2010; 31:209-19. [PMID: 19866474 DOI: 10.1002/bem.20550] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Osteogenesis is a complex series of events involving the differentiation of mesenchymal stem cells to generate new bone. In this study, we examined the effect of pulsed electromagnetic fields (PEMFs) on cell proliferation, alkaline phosphatase (ALP) activity, mineralization of the extracellular matrix, and gene expression in bone marrow mesenchymal stem cells (BMMSCs) during osteogenic differentiation. Exposure of BMMSCs to PEMFs increased cell proliferation by 29.6% compared to untreated cells at day 1 of differentiation. Semi-quantitative RT-PCR indicated that PEMFs significantly altered temporal expression of osteogenesis-related genes, including a 2.7-fold increase in expression of the key osteogenesis regulatory gene cbfa1, compared to untreated controls. In addition, exposure to PEMFs significantly increased ALP expression during the early stages of osteogenesis and substantially enhanced mineralization near the midpoint of osteogenesis. These results suggest that PEMFs enhance early cell proliferation in BMMSC-mediated osteogenesis, and accelerate the osteogenesis.
Collapse
Affiliation(s)
- Li-Yi Sun
- Department of Biological Science and Technology, National Chiao Tung University, No. 75 Po-Ai Street, Hsinchu, Taiwan, ROC
| | | | | | | | | |
Collapse
|
50
|
Ng SY, Chin CH, Lau YT, Luo J, Wong CK, Bian ZX, Tsang SY. Role of voltage-gated potassium channels in the fate determination of embryonic stem cells. J Cell Physiol 2010; 224:165-77. [PMID: 20333647 DOI: 10.1002/jcp.22113] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Embryonic stem cells (ESCs) possess two unique characteristics: self-renewal and pluripotency. In this study, roles of voltage-gated potassium channels (K(v)) in maintaining mouse (m) ESC characteristics were investigated. Tetraethylammonium (TEA(+)), a K(v) blocker, attenuated cell proliferation in a concentration-dependent manner. Possible reasons for this attenuation, including cytotoxicity, cell cycle arrest and differentiation, were examined. Blocking K(v) did not change the viability of mESCs. Interestingly, K(v) inhibition increased the proportion of cells in G(0)/G(1) phase and decreased that in S phase. This change in cell cycle distribution can be attributed to cell cycle arrest or differentiation. Loss of pluripotency as determined at both molecular and functional levels was detected in mESCs with K(v) blockade, indicating that K(v) inhibition in undifferentiated mESCs directs cells to differentiate instead of to self-renew and progress through the cell cycle. Membrane potential measurement revealed that K(v) blockade led to depolarization, consistent with the role of K(v) as the key determinant of membrane potential. The present results suggest that membrane potential changes may act as a "switch" for ESCs to decide whether to proliferate or to differentiate: hyperpolarization at G(1) phase would favor ESCs to enter S phase while depolarization would favor ESCs to differentiate. Consistent with this notion, S-phase-synchronized mESCs were found to be more hyperpolarized than G(0)/G(1)-phase-synchronized mESCs. Moreover, when mESCs differentiated, the differentiation derivatives depolarized at the initial stage of differentiation. This investigation is the first study to provide evidence that K(v) and membrane potential affect the fate determination of ESCs.
Collapse
Affiliation(s)
- Sze-Ying Ng
- Department of Biochemistry, The Chinese University of Hong Kong, Hong Kong, China
| | | | | | | | | | | | | |
Collapse
|